Aging and drug discovery

3 downloads 0 Views 362KB Size Report
Nov 13, 2018 - Department of Physiology, Perelman School of Medicine, University of Pennsylvania, .... (Copenhagen, Denmark) discussed the phenotypical.
www.aging‐us.com 

 

 

         AGING 2018, Vol. 10, Advance Meeting Report

Aging and drug discovery   

Daniela  Bakula1,  Alexander  M.  Aliper2,  Polina  Mamoshina2,  Michael  A.  Petr1,  Amanuel  Teklu1, Joseph  A.  Baur3,4,  Judith  Campisi5,6,  Collin  Y.  Ewald7,  Anastasia  Georgievskaya8,  Vadim  N. Gladyshev9,10,  Olga  Kovalchuk11,12,  Dudley  W.  Lamming13,  Martijn  S.  Luijsterburg14,  Alejandro Martín‐Montalvo15,  Stuart  Maudsley16,17,  Garik  V.  Mkrtchyan1,  Alexey  Moskalev18,19,20,  S.  Jay Olshansky21, Ivan V. Ozerov2, Alexander Pickett22, Michael Ristow7, Alex Zhavoronkov2, Morten Scheibye‐Knudsen1*   

1

Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen,  Copenhagen, Denmark   2 Pharmaceutical Artificial Intelligence Department, Insilico Medicine, Inc., Baltimore, MD 20850, USA  3 Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania,  Philadelphia, PA 19104, USA   4 Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA  5 Buck Institute for Research on Aging, Novato, CA 94945, USA   6 Lawrence Berkeley National Laboratory, Life Sciences Division, Berkeley, CA 94720, USA   7 Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute for  Technology (ETH) Zürich, Switzerland   8 Youth Laboratories, Moscow, Russia   9 Division of Genetics, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston,  MA 02115, USA   10 Belozersky Institute of Physico‐Chemical Biology, Moscow State University, Moscow, Russia   11 Department of Biological Sciences, University of Lethbridge, Lethbridge, Alberta, Canada   12 Pathway Rx Ltd, Lethbridge, Alberta, Canada   13 Department of Medicine, University of Wisconsin‐Madison, Madison, WI 53706, USA   14 Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands   15 Pancreatic Islet Development and Regeneration Unit/Laboratory of Aging Biology, Centro Andaluz de Biologı́a  Molecular y Medicina Regenerativa‐CABIMER, Universidad de Sevilla‐CSIC‐Universidad Pablo de Olavide, Seville,  Spain   16 Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium   17 Translational Neurobiology Group, Center of Molecular Neurology, VIB, Antwerp, Belgium   18 Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia   19 Institute of Biology of Komi Science Center of Ural Branch of RAS, Syktyvkar, Russia   20 Moscow Institute of Physics and Technology, Dolgoprudny, Russia   21 Division of Epidemiology and Biostatistics, University of Illinois at Chicago School of Public Health, Chicago, IL  60612 USA   22 Juvenescence Limited, Douglas, Isle of Man, UK     Correspondence to: Morten Scheibye‐Knudsen; email:  [email protected]  Keywords: aging, drug discovery  Received:  October 8, 2018  Accepted:  November 4, 2018  Published:  November 13, 2018    Copyright:  Bakula  et  al.  This  is  an  open‐access  article  distributed  under  the  terms  of  the  Creative  Commons  Attribution License  (CC  BY  3.0),  which  permits  unrestricted  use,  distribution,  and  reproduction  in  any  medium,  provided  the  original author and source are credited.

www.aging‐us.com 



AGING

ABSTRACT Multiple interventions in the aging process have been discovered to extend the healthspan of model organisms. Both  industry  and  academia  are  therefore  exploring  possible  transformative  molecules  that  target  aging  and age‐associated diseases. In this overview, we summarize the presented talks and discussion points of the 5th Annual  Aging  and  Drug  Discovery  Forum  2018  in  Basel,  Switzerland.  Here  academia  and  industry  came together,  to  discuss  the  latest  progress  and  issues  in  aging  research.  The  meeting  covered  talks  about  the mechanistic cause of aging, how longevity signatures may be highly conserved, emerging biomarkers of aging, possible  interventions  in  the  aging  process  and  the  use  of  artificial  intelligence  for  aging  research  and  drug discovery.  Importantly,  a  consensus  is  emerging  both  in  industry  and  academia,  that  molecules  able  to intervene in the aging process may contain the potential to transform both societies and healthcare. 

INTRODUCTION

represents a double-edged sword by repressing malignant transformation. However, accumulation over time causes degenerative phenotypes and drives tumor formation. Therefore, targeting senescent cells using senolytics or by compensating for the beneficial effects of the induced secretory phenotype, may help to prevent age-dependent functional decline. Clear markers for senescent cells are still lacking, due to the strong heterogeneity of senescent cells [3,4]. To address this issue, the group of Judith Campisi implemented single cell analysis to further understand the cell-type specific signatures of senescent cells, which may help to find more specific targets [4].

“Why do we age?”; “Can we intervene in the aging process?”; and if so, what approaches should aging science take to transform research into viable therapeutic interventions to improve public health? Understanding the mechanisms of aging will be of vital importance to answering these questions. However, several obstacles stand in the way of generating efficacious and safe interventions that extend the period of healthy life. At the 5th Annual Aging and Drug Discovery Forum which was held during the Basel Life Congress, Basel, Switzerland, September 12-13, 2018, leading aging experts from academia and industry came together to discuss top issues in aging research. The meeting was organized by Alex Zhavoronkov, CEO of Insilico Medicine (Baltimore, MD, United States), a company specialized in implementing artificial intelligence (AI) solutions for drug discovery and biomarker development and Morten Scheibye-Knudsen, head of the aging interventions lab at the Center for Healthy Aging, University of Copenhagen (Copenhagen, Denmark). Here, we provide a brief overview of the presented results and discussion points.

Michael Ristow from the ETH Zürich (Zürich, Switzerland) presented his work about redox-regulation of lifespan and metabolism. Previous results of his group suggest the concept of mitohormesis as a healthpromoting signaling pathway, where low concentrations of reactive oxygen species (ROS) lead to an adaptive response [5]. The concept of mitohormesis was further supported by the observation that glucose restriction in C. elegans leads to increased ROS production, thereby promoting endogenous stress defense and increased lifespan [6]. ROS-induced increased insulin sensitivity was also observed in a physical exercise trial in humans and, interestingly, this effect was inhibited by the supplementation of antioxidants [7]. Furthermore, his group compared gene expression levels of young, mature and old C. elegans, D. rerio and M. musculus to predict evolutionary conserved age-related genes. Branched-chain amino acid transferase 1 (bcat-1) was identified as a strong lifespan regulator and, accordingly, knockdown of bcat-1 increased lifespan in C. elegans [8].

What drives the aging process? Aging is a multifactorial process that leads to loss of cellular homeostasis followed by susceptibility to diseases. The functional decline is influenced by highly conserved signaling pathways that could be targeted to manipulate the aging process. Judith Campisi from the Buck Institute for Research on Aging (Novato, CA, United States) highlighted the impact of cellular senescence as an evolutionary balancing act on aging [1]. Senescent cells accumulate over time and promote the aging processes by loss of tissue functionality and through the secretion of pro-inflammatory factors, known as the secretory phenotype [2]. Senescence

www.aging‐us.com 

Both endogenous stress factors, such as ROS and exogenous stress factors are constantly challenging the human genome. Different DNA repair pathways have evolved to repair the different DNA lesions. Indeed, DNA damage accumulate with age and DNA repair



AGING

capacity has been suggested to decline with age [9]. The impact of DNA repair on aging was emphasized by two presentations at the meeting. Morten ScheibyeKnudsen from the University of Copenhagen (Copenhagen, Denmark) discussed the phenotypical landscape of aging [10] and emphasized that the mechanisms causing the different pathologies of aging are poorly understood. Morten Scheibye-Knudsen presented the value of using hierarchical clustering and machine learning algorithms to compare similarities between diseases based on their clinical features [11]. This approach may help to find cellular pathways that are linked to specific disease pathologies. Notably, diseases caused by defects in DNA repair resembles many features of aging, emphasizing genome maintenance as a key factor of aging [12]. Thus, premature aging disorders may be good model systems to study aging.

showed that caloric restriction, as well as the long-lived mutant Snell dwarf mice, exhibit slower epigenetic aging. A further study of his group determined longevity-related transcriptomic changes across 33 species of mammals [20] and 14 different Drosophila species [21], which may be used for prediction of new longevity interventions. Collin Ewald from the ETH Zürich (Zürich, Switzerland) presented his recent work on the transcriptomic signatures of longevity in C. elegans, revealing that the extracellular matrix (ECM) composition undergo changes with age. Notably, potential aging interventions, such as rapamycin treatment, modulate ECM gene expression [22]. Moreover, reduced insulin/IGF-1 signaling induced lifespan extension was shown to be dependent on collagen gene expression. These observations indicate that modulation of the ECM may be a promising target. Collin Ewald’s work reveals the value of C. elegans as a powerful model system to study aging and aging interventions, due to its relatively short life span and easily manipulated genome.

This observation was underscored by Martijn Luijsterburg from the Leiden University Medical Center (Leiden, The Netherlands) who presented his latest research on transcription-coupled DNA repair, a DNA repair pathway that removes lesions blocking transcription of active genes. Interestingly, mutations in transcription-coupled repair genes are known to cause diverse phenotypes [13]. Mutations in CSA and CSB can cause the premature aging disorder Cockayne syndrome which is characterized by severe neurodegeneration, whereas mutations in UVSSA can cause the UV-sensitivity syndrome, a disease without neurological phenotypes. Untangling the precise molecular functions of the transcription coupled repair complex members may help to understand the disease mechanism of Cockayne syndrome and aging features. Martijn Luijsterburg presented his latest results towards this understanding.

In addition to epigenetic and gene expression changes, profound age-dependent remodeling of protein expression can be observed with age [23]. Stuart Maudsley from the University of Antwerp (Antwerp, Belgium) discussed the identification of multidimensional regulators of aging. In several recent studies, his group has examined, using a combination of classical signaling pathway analysis and natural language processing informatics, age-related proteomic alterations in the hypothalamus, the potential key organ that coordinates global somatic aging [24]. Using this novel approach, the G protein-coupled receptor kinase interacting protein 2 (GIT2) was identified as a potential key regulator of aging. Consequently, GIT2 protein expression was demonstrated to be strongly agesensitive in the hypothalamus and multiple other brain regions as well as a broad range of peripheral tissues associated with energy metabolism regulation [24,25]. GIT2 acts a scaffold protein for multiple signaling pathways, hence decreased GIT2 function affects several hallmarks of aging including metabolic dysfunction [25], DNA damage responses [26], oxidative damage responsiveness [27] and immune system senescence [28]. Thus, GIT2 may be a promising multidimensional therapeutic target for age-associated diseases [29,30].

Signatures of longevity In recent years, a concerted effort was made to identify biological markers that can predict the chronological age of an individual. Different types of age predictors including telomere length [14], gene expression changes [15] and epigenetic changes, the latter known as the epigenetic clock [16–18], have been shown to strongly correlate with chronological aging. Vadim Gladyshev from the Brigham and Women’s Hospital, Harvard Medical School (Boston, MA, United States) and Moscow State University (Moscow, Russia) highlighted that the combination of different approaches can allow for more reliable biological age prediction, since aging is a systemic process. His research group investigated the mouse blood DNA methylome of different age groups to develop an epigenetic clock that can be used to test aging interventions [19]. Notably, this analysis

www.aging‐us.com 

Interventions in aging and aging-associated disease In the last years multiple interventions for life span extension have been identified. Pharmacological (e.g. rapamycin, resveratrol, metformin) and non-pharma-



AGING

cological interventions (e.g. caloric restriction, physical exercise) have been extensively discussed. Nevertheless, how these interventions may affect human health span is not well understood. A growing number of studies suggest that the macronutrient composition of the diet, and in particular dietary protein, may also play a crucial role in regulating metabolic health and life span [31]. However, it remains an open question as whether altered consumption of specific dietary amino acids mediates the beneficial effects of reduced dietary protein. In this regard, Dudley Lamming from the University of Wisconsin (Madison, WI, United States) presented his work about promoting healthy aging through the reduction of specific dietary macronutrients. Dudley Lamming’s research group provided data revealing that the reduction of branched chain amino acids improved metabolic health in lean as well as dietinduced obese C57BL/6J mice, and recapitulated many of the metabolic benefits of a low protein diet [32,33]. Notably, a reduction of leucine alone stimulated white adipose tissue growth, indicating that individual branched-chain amino acids may have distinct roles in the regulation of metabolic health [33].

increased the median lifespan of C. elegans and D. melanogaster, along with changes associated with aging such as increased stress resistance. At the molecular level the response to fucoxanthin in D. melanogaster goes along with transcriptional changes of longevityrelated signaling pathways, such as the MAPK, mTOR and autophagy pathways [39]. Preliminary data on human fibroblasts shows antisenogenic effects of fucoxanthin at 100 nM and 1 nkM (unpublished). Olga Kovalchuk from the University of Lethbridge and PathwayRx Inc. (Lethbridge, Canada) gave an overview of skin aging driven by intrinsic and extrinsic factors leading to deregulated matrix metalloproteinases (MMPs), enzymes activated by UV exposure, inflammation and other factors [40]. MMPs contribute to the breakdown of collagen while inhibiting new collagen formation, and cytokine deregulation could be possible intervention targets by novel extracts. She gave an overview of the effects of UV and THz radiation on skin, and presented novel (patent pending) plant extracts with very potent anti-inflammatory and anti-aging capacities that can be used to counteract harmful environmental effects of skin and potentially reverse skin aging.

Due to the difficulty to reduce caloric intake, compounds mimicking the positive effects of caloric restriction may be a promising strategy, as outlined by Joseph Baur from the University of Pennsylvania (Philadelphia, PA, United States). Modulating nicotinamide adenine dinucleotide (NAD+) levels is a promising strategy to treat age-associated physiological decline [34,35]. Recent work from Joseph Baur revealed that muscle specific knockout of nicotinamide phosphoribosyltransferase (NAMPT), an enzyme that is essential for the maintenance of normal NAD+ concentration, lead to progressive loss of muscle function. The observed phenotype was counteracted by administration of the NAD+ precursor nicotinamide riboside [36]. Interestingly, lifelong NAMPT overexpression prevented the age-related loss of NAD+ and improved exercise performance in old mice. Although mitochondrial NAD+ pools have been shown to contribute to cell metabolism, the source of mitochondrial NAD+ remains an obscurity. A recently published study by the group of Joseph Baur proposes the existence of a so far undiscovered mitochondrial NAD+ transporter, which may be a further potential target to modulate the compartmentalization of NAD+ within the cell [37].

Alejandro Martín-Montalvo from the Andalusian Molecular Biology and Regenerative Medicine Centre CABIMER- (Seville, Spain) discussed the potential of interventions based on the use of thyroid hormones on aging and age-associated diseases. The key role of thyroid hormone levels in regulating longevity is corroborated by the observation, that humans with exceptional longevity, and other long-lived animals, show decreased circulating thyroid hormone levels [41]. Nevertheless, Alejandro Martín-Montalvo showed that thyroid hormones enhance glucose clearance, leading him to investigate if these hormones could be used for the treatment of different types of diabetes. Indeed, his team has shown, using two experimental models of diabetes, that thyroid hormones efficiently blunt the onset of diabetes and increase survival in mice [42]. The data indicate a potential benefit of thyroid hormones and/or thyromimetics for the treatment of type 1 diabetes mellitus. The power of AI to facilitate anti-aging drug discovery In comparison to conventional drug development, the development and commercialization of pharmaceuticals targeting aging pose additional difficulties. Alexander Pickett (Juvenescence Ltd, Boston, MA, United States), SVP of Business Development at Juvenescence Limited, a drug development company focusing on therapeutics that modify aging, highlighted the

Alexey Moskalev from the Moscow Institute of Physics and Technology (Moscow, Russia) emphasized the potential of natural compounds as aging interventions. Alexey Moskalev presented data about a potential geroprotective function of the carotenoid fucoxanthin in C. elegans and D. melanogaster [38]. Fucoxanthin

www.aging‐us.com 



AGING

obstacles to developing aging interventions. The lack of reliable techniques to prove the efficacy of the therapeutics is one of the main difficulties of clinical trial designs for aging interventions. In this regard, Alexander Pickett pointed out the importance of developing new reliable biomarkers and validating that they were not only predictive of aging but also responded to known interventions. Another obstacle will be commercializing aging intervention drugs: so far, our health care systems rely predominantly on treating illness rather than keeping people healthy. Juvenescence believes that developing aging interventions to treat existing diseases and seeking outcomes-based reimbursement is the quickest path to bringing aging interventions to large populations. To address these issues, Juvenescence Limited has invested in several biotech startups that pursue a whole range of approaches, including Insilico Medicine.

revealed, which makes it relevant to insurance and financial sectors as well. Facial analytics as a promising biomarker for aging, was further acknowledged by Anastasia Georgievskaya, a co-founder of Youth Laboratories (Moscow, Russian Federation). Youth Laboratories is using artificial intelligence to study aging and discover new class of non-invasive photographic aging biomarkers. Currently, the company is collecting photographs of lab mice to develop the first photographic biomarker for mice. Since mice are still the main model of aging research, the photographic mouse aging clock may facilitate the identification of beneficial aging interventions in mice to further translate them to humans. Panel discussion on key trends in longevity biotechnology The panel discussion on longevity biotechnology industry with Vadim N. Gladyshev, Morten ScheibyeKnudsen, Judith Campisi, Alexander Pickett, Michael Antonov, Joseph A. Baur and Stuart Maudsley and chaired by Alex Zhavoronkov, identified the following areas with the most potential of commercialization: repurposing of drugs that are already on the market, senolytics, mTOR inhibitors, NAD+ activators and modulators, reprogramming-based and regenerativebased approaches, AI and data analysis, and digital health.

Ivan Ozerov from Insilico Medicine (Baltimore, MD, United States) introduced an AI-driven computational pipeline approach to identify new small molecules that counteract the progression of cellular senescence. The approach relies on the concept of 5R (Rescue, Remove, Replenish, Reinforce, Repeat) and utilize the recently published in silico Pathway Activation Network Decomposition Analysis (iPANDA) method [43]. The pipeline may accelerate target and drug discovery for anti-aging interventions.

During the discussion, the panel highlighted the great potential of drug repurposing (repositioning) for aging research. Drug repurposing or target extension allows the identification of new indications for drugs with low safety risk profiles, as they have been already tested in humans. Great progress has been made in developing approaches for drug characterization and classification that could be used for drug repurposing [45–47]. Indeed, several attempts have been made to predict a potential aging application of existing medications [48– 52].

The power of Artificial Intelligence to facilitate drug discovery was highlighted further by Garik Mkrtchyan from the Scheibye-Knudsen lab (Copenhagen University, Denmark). Increased unrepaired DNA lesions are a hallmark of aging. To counteract this, stimulating DNA repair may be a promising approach. Garik Mkrtchyan presented work about deep learning algorithms utilized to screen a small molecule library to identify compounds that stimulate DNA repair. The success of the approach was proven by the identification of drugs that induced cellular resistance to ionizing radiation but do not cause DNA damage (unpublished data).

Recently, several senolytics, selective inductors of senescence cell death, have been tested as aging preventive or aging revertive agents [53,54]. At the same time, the panel emphasized that such therapies have high-risk profiles, as they are only in early phases of clinical development [55]. mTOR inhibitors [56] and NAD+ activators [57,58] have also been listed as interventions that could delay the onset of multiple agerelated pathologies. Therefore, they could be explored as human aging therapies in the nearest future. One panelist made the point that regenerative medicine approaches [59] could provide therapeutic possibilities for targeting aging and could be developed further to reach the market.

S. Jay Olshansky from the University of Illinois (Chicago) and co-founder of Lapetus Solutions, Inc., further emphasized the requirement of biomarkers to facilitate the identification of aging interventions. He made a point that the target of intervention should be healthspan and not lifespan [44]. Olshansky showed that biological age is reflected in facial features and underlined the value of photographic phenotypic biomarkers in health assessment. Based on facial photographs, age and gender can be predicted with a high degree of reliability, but also further risk factors such as smoking behavior and body mass index can be

www.aging‐us.com 



AGING

AI and deep learning are now recognized as transformative technologies in healthcare in general [47,60,61]. For instance, a lot of progress has been made in machine-learning-based biomarkers of human aging using easily obtained data such as methylation [16,17], transcriptomics [62], proteomics [63] and blood biochemistry [64,65]. Following an extensive discussion, panelists selected AI and digital health as key developments that would catalyze the pace of innovation in aging research. All panelists agreed that aging research and longevity interventions have tremendous opportunities to impact the whole healthcare sector.

University of Wisconsin-Madison School of Medicine and Public Health and Department of Medicine, as well as the facilities and resources of the William S. Middleton Memorial Veterans Hospital. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. This work does not represent the views of the Department of Veterans Affairs or the United States Government. MSL is supported by an LUMC research fellowship and a VIDI grant from the Netherlands scientific organization (NWO- ALW-016.161.320). A.M.-M. is supported by grants from the Instituto de Salud Carlos III co-funded by Instituto de Salud Carlos III and FEdeR (CP14/00105 and PI15/00134). SM was supported by the FWO-OP/Odysseus program (42/FA010100/32/6484). SJO's current work is funded by The Glenn Award from the Glenn Foundation for Medical Research. MR is supported by the Swiss National Science Foundation and the European Union Horizon 2020 program. MSK is supported by grants from the Danish Cancer Society (#R167-A11015_001), the Independent Research Fund Denmark (#701600230B) and the Novo Nordisk Foundation (NNF17OC0027812).

Closing remarks Clearly, aging interventions require a multifaceted approach through which complex phenotypes can be evaluated using next generation machine learning approaches. The meeting underscored the awareness that drug discovery in the aging field will require not only determination from academic researchers but also multiple industrial partners and investors. This state of the field is particularly clear in light of the significant number of emerging companies that attempt to tackle the task of finding translatable interventions into aging. It is evident from the formation of all these startups that the next promising longevity pharmaceuticals will be found. The future looks bright.

REFERENCES 1.   Campisi J, Robert L. Cell senescence: role in aging and  age‐related diseases.  Interdiscip  Top  Gerontol.  2014;  39:45–61. https://doi.org/10.1159/000358899 

ACKNOWLEDGEMENTS

2.   Coppé  JP,  Desprez  PY,  Krtolica  A,  Campisi  J.  The  senescence‐associated secretory phenotype: the dark  side  of  tumor  suppression.  Annu  Rev  Pathol.  2010;  5:99–118.  https://doi.org/10.1146/annurev‐pathol‐ 121808‐102144 

We would like to thank the entire team behind Basel Life for their strong and dedicated help in all aspects of this meeting. In particular, we would like to thank Krisztina Németh and Bhupinder Bhullar for their great support for our program. We would also like to thank Insilico Medicine and Aging-US for their support of the meeting.

3.   Hernandez‐Segura A, de Jong TV, Melov S, Guryev V,  Campisi  J,  Demaria  M.  Unmasking  Transcriptional  Heterogeneity  in  Senescent  Cells.  Curr  Biol.  2017;  27:2652–2660.e4.  https://doi.org/10.1016/j.cub.2017.07.033 

CONFLICTS OF INTEREST The authors declare no conflicts of interest.

4.   Wiley CD, Flynn JM, Morrissey C, Lebofsky R, Shuga J,  Dong X, Unger MA, Vijg J, Melov S, Campisi J. Analysis  of  individual  cells  identifies  cell‐to‐cell  variability  following induction of cellular senescence. Aging Cell.  2017; 16:1043–50.     https://doi.org/10.1111/acel.12632 

FUNDING DB is supported by the German Research Foundation (Forschungsstipendium; BA 6276/1-1). CYE is supported by Swiss National Science Foundation [163898]. VNG is supported by grants from National Institutes of Health, and by the Russian Federation grant 14.W03.31.0012. DWL presented the results of research supported in part by research grants and funds from the National Institutes of Health, the Wisconsin Partnership Program, the Progeria Research Foundation, the American Federation for Aging Research, and the

www.aging‐us.com 

5.   Ristow  M.  Unraveling  the  truth  about  antioxidants:  mitohormesis  explains  ROS‐induced  health  benefits.  Nat Med. 2014; 20:709–11.     https://doi.org/10.1038/nm.3624  6.   Schulz  TJ,  Zarse  K,  Voigt  A,  Urban  N,  Birringer  M,  Ristow M. Glucose restriction extends Caenorhabditis  elegans  life  span  by  inducing  mitochondrial 



AGING

 

and  cell  types.  Genome  Biol.  2013;  14:R115.  https://doi.org/10.1186/gb‐2013‐14‐10‐r115 

respiration  and  increasing  oxidative  stress.  Cell  Metab. 2007; 6:280–93.   https://doi.org/10.1016/j.cmet.2007.08.011 

18.  Weidner CI, Lin Q, Koch CM, Eisele L, Beier F, Ziegler  P, Bauerschlag DO, Jöckel KH, Erbel R, Mühleisen TW,  Zenke  M,  Brümmendorf  TH,  Wagner  W.  Aging  of  blood can be tracked by DNA methylation changes at  just  three  CpG  sites.  Genome  Biol.  2014;  15:R24.  https://doi.org/10.1186/gb‐2014‐15‐2‐r24 

7.   Ristow  M,  Zarse  K,  Oberbach  A,  Klöting  N,  Birringer  M,  Kiehntopf  M,  Stumvoll  M,  Kahn  CR,  Blüher  M.  Antioxidants  prevent  health‐promoting  effects  of  physical exercise in humans. Proc Natl Acad Sci USA.  2009; 106:8665–70.     https://doi.org/10.1073/pnas.0903485106 

19.  Petkovich  DA,  Podolskiy  DI,  Lobanov  AV,  Lee  SG,  Miller  RA,  Gladyshev  VN.  Using  DNA  Methylation  Profiling  to  Evaluate  Biological  Age  and  Longevity  Interventions.  Cell  Metab.  2017;  25:954–960.e6.  https://doi.org/10.1016/j.cmet.2017.03.016 

8.   Mansfeld  J,  Urban  N,  Priebe  S,  Groth  M,  Frahm  C,  Hartmann  N,  Gebauer  J,  Ravichandran  M,  Dommaschk  A,  Schmeisser  S,  Kuhlow  D,  Monajembashi  S,  Bremer‐Streck  S,  et  al.  Branched‐ chain amino acid catabolism is a conserved regulator  of physiological ageing. Nat Commun. 2015; 6:10043.  https://doi.org/10.1038/ncomms10043 

20.  Fushan AA, Turanov AA, Lee SG, Kim EB, Lobanov AV,  Yim SH, Buffenstein R, Lee SR, Chang KT, Rhee H, Kim  JS,  Yang  KS,  Gladyshev  VN.  Gene  expression  defines  natural  changes  in  mammalian  lifespan.  Aging  Cell.  2015; 14:352–65. https://doi.org/10.1111/acel.12283 

9.   Vijg  J,  Dong  X,  Milholland  B,  Zhang  L.  Genome  instability: a conserved mechanism of ageing? Essays  Biochem. 2017; 61:305–15.     https://doi.org/10.1042/EBC20160082  10.  Scheibye‐Knudsen  M.  Neurodegeneration  accelerated aging. Dan Med J. 2016; 63:63. 

21.  Ma  S,  Avanesov  AS,  Porter  E,  Lee  BC,  Mariotti  M,  Zemskaya  N,  Guigo  R,  Moskalev  AA,  Gladyshev  VN.  Comparative  transcriptomics  across  14  Drosophila  species  reveals  signatures  of  longevity.  Aging  Cell.  2018; 17:e12740. https://doi.org/10.1111/acel.12740 

in 

11.  Scheibye‐Knudsen  M,  Scheibye‐Alsing  K,  Canugovi  C,  Croteau DL, Bohr  VA. A  novel  diagnostic  tool  reveals  mitochondrial  pathology  in  human  diseases  and  aging.  Aging  (Albany  NY).  2013;  5:192–208.  https://doi.org/10.18632/aging.100546 

22.  Ewald  CY,  Landis  JN,  Porter  Abate  J,  Murphy  CT,  Blackwell  TK.  Dauer‐independent  insulin/IGF‐1‐ signalling  implicates  collagen  remodelling  in  longevity. Nature. 2015; 519:97–101.     https://doi.org/10.1038/nature14021 

12.  Keijzers  G,  Bakula  D,  Scheibye‐Knudsen  M.  Monogenic  Diseases  of  DNA  Repair.  N  Engl  J  Med.  2017; 377:1868–76.     https://doi.org/10.1056/NEJMra1703366 

23.  Walther DM, Kasturi P, Zheng M, Pinkert S, Vecchi G,  Ciryam P, Morimoto RI, Dobson CM, Vendruscolo M,  Mann  M,  Hartl  FU.  Widespread  Proteome  Remodeling and Aggregation in Aging C. elegans. Cell.  2015; 161:919–32.     https://doi.org/10.1016/j.cell.2015.03.032 

13.  Marteijn  JA,  Lans  H,  Vermeulen  W,  Hoeijmakers  JH.  Understanding nucleotide excision repair and its roles  in  cancer  and  ageing.  Nat  Rev  Mol  Cell  Biol.  2014;  15:465–81. https://doi.org/10.1038/nrm3822 

24.  Chadwick W, Martin B, Chapter MC, Park SS, Wang L,  Daimon CM, Brenneman R, Maudsley S. GIT2 acts as a  potential keystone protein in functional hypothalamic  networks  associated  with  age‐related  phenotypic  changes in rats. PLoS One. 2012; 7:e36975.     https://doi.org/10.1371/journal.pone.0036975 

14.  Mather  KA,  Jorm  AF,  Parslow  RA,  Christensen  H.  Is  telomere  length  a  biomarker  of  aging?  A  review.  J  Gerontol  A  Biol  Sci  Med  Sci.  2011;  66:202–13.  https://doi.org/10.1093/gerona/glq180  15. de Magalhães JP, Curado J, Church GM. Meta‐analysis  of  age‐related  gene  expression  profiles  identifies  common  signatures  of  aging.  Bioinformatics.  2009;  25:875–81.  https://doi.org/10.1093/bioinformatics/btp073 

25.  Martin  B,  Chadwick  W,  Janssens  J,  Premont  RT,  Schmalzigaug  R,  Becker  KG,  Lehrmann  E,  Wood  WH,  Zhang Y, Siddiqui S, Park SS. Cong W, Daimon CM, et  al.  GIT2  Acts  as  a  Systems‐Level  Coordinator  of  Neurometabolic  Activity  and  Pathophysiological  Aging. Front Endocrinol. 2016; 6:191.     https://doi.org/10.3389/fendo.2015.00191 

16.  Hannum  G,  Guinney  J,  Zhao  L,  Zhang  L,  Hughes  G,  Sadda  S,  Klotzle  B,  Bibikova  M,  Fan  JB,  Gao  Y,  Deconde R, Chen M, Rajapakse I, et al. Genome‐wide  methylation  profiles  reveal  quantitative  views  of  human  aging  rates.  Mol  Cell.  2013;  49:359–67.  https://doi.org/10.1016/j.molcel.2012.10.016 

26.  Lu  D,  Cai  H,  Park  SS,  Siddiqui  S,  Premont  RT,  Schmalzigaug  R,  Paramasivam  M,  Seidman  M,  Bodogai I, Biragyn A, Daimon CM, Martin B, Maudsley  S.  Nuclear  GIT2  is  an  ATM  substrate  and  promotes  DNA repair. Mol Cell Biol. 2015; 35:1081–96.  

17.  Horvath S. DNA methylation age of human tissues  

www.aging‐us.com 



AGING

https://doi.org/10.1128/MCB.01432‐14 

36.  Frederick DW, Loro E, Liu L, Davila A Jr, Chellappa K,  Silverman IM, Quinn WJ 3rd, Gosai SJ, Tichy ED, Davis  JG, Mourkioti F, Gregory BD, Dellinger RW, et al. Loss  of  NAD  Homeostasis  Leads  to  Progressive  and  Reversible  Degeneration  of  Skeletal  Muscle.  Cell  Metab. 2016; 24:269–82.     https://doi.org/10.1016/j.cmet.2016.07.005 

27.  Chadwick  W,  Zhou  Y,  Park  SS,  Wang  L,  Mitchell  N,  Stone MD, Becker KG, Martin B, Maudsley S. Minimal  peroxide  exposure  of  neuronal  cells  induces  multifaceted  adaptive  responses.  PLoS  One.  2010;  5:e14352.  https://doi.org/10.1371/journal.pone.0014352 

37.  Davila  A,  Liu  L,  Chellappa  K,  Redpath  P,  Nakamaru‐ Ogiso E, Paolella LM, Zhang Z, Migaud ME, Rabinowitz  JD,  Baur  JA.  Nicotinamide  adenine  dinucleotide  is  transported  into  mammalian  mitochondria.  eLife.  2018; 7:7. https://doi.org/10.7554/eLife.33246 

28.  Siddiqui S, Lustig A, Carter A, Sankar M, Daimon CM,  Premont RT, Etienne H, van Gastel J, Azmi A, Janssens  J,  Becker  KG,  Zhang  Y,  Wood  W,  et  al.  Genomic  deletion  of  GIT2  induces  a  premature  age‐related  thymic  dysfunction  and  systemic  immune  system  disruption.  Aging  (Albany  NY).  2017;  9:706–40.  https://doi.org/10.18632/aging.101185 

38.  Lashmanova  E,  Proshkina  E,  Zhikrivetskaya  S,  Shevchenko  O,  Marusich  E,  Leonov  S,  Melerzanov  A,  Zhavoronkov  A,  Moskalev  A.  Fucoxanthin  increases  lifespan  of  Drosophila  melanogaster  and  Caenorhabditis  elegans.  Pharmacol  Res.  2015;  100:228–41.  https://doi.org/10.1016/j.phrs.2015.08.009 

29.  van  Gastel  J,  Boddaert  J,  Jushaj  A,  Premont  RT,  Luttrell LM, Janssens J, Martin B, Maudsley S. GIT2‐A  keystone  in  ageing  and  age‐related  disease.  Ageing  Res Rev. 2018; 43:46–63.     https://doi.org/10.1016/j.arr.2018.02.002 

39.  Moskalev  A,  Shaposhnikov  M,  Zemskaya  N,  Belyi  A,  Dobrovolskaya E, Patova A, Guvatova Z, Lukyanova E,  Snezhkina  A,  Kudryavtseva  A.  Transcriptome  analysis  reveals  mechanisms  of  geroprotective  effects  of  fucoxanthin  in  Drosophila.  BMC  Genomics.  2018  (Suppl  3);  19:77.  https://doi.org/10.1186/s12864‐ 018‐4471‐x 

30.  Leysen  H,  van  Gastel  J,  Hendrickx  JO,  Santos‐Otte  P,  Martin  B,  Maudsley  S.  G  Protein‐Coupled  Receptor  Systems  as  Crucial  Regulators  of  DNA  Damage  Response  Processes.  Int  J  Mol  Sci.  2018;  19:19.  https://doi.org/10.3390/ijms19102919  31.  Solon‐Biet  SM,  McMahon  AC,  Ballard  JW,  Ruohonen  K, Wu LE, Cogger VC, Warren A, Huang X, Pichaud N,  Melvin  RG,  Gokarn  R,  Khalil  M,  Turner  N,  et  al.  The  ratio  of  macronutrients,  not  caloric  intake,  dictates  cardiometabolic  health,  aging,  and  longevity  in  ad  libitum‐fed  mice.  Cell  Metab.  2014;  19:418–30.  https://doi.org/10.1016/j.cmet.2014.02.009 

40.  Krutmann  J,  Bouloc  A,  Sore  G,  Bernard  BA,  Passeron  T.  The  skin  aging  exposome.  J  Dermatol  Sci.  2017;  85:152–61.  https://doi.org/10.1016/j.jdermsci.2016.09.015  41.  Barzilai N, Huffman DM, Muzumdar RH, Bartke A. The  critical role of metabolic pathways in aging. Diabetes.  2012;  61:1315–22.  https://doi.org/10.2337/db11‐ 1300 

32.  Cummings NE, Williams EM, Kasza I, Konon EN, Schaid  MD, Schmidt BA, Poudel C, Sherman DS, Yu D, Arriola  Apelo  SI,  Cottrell  SE,  Geiger  G,  Barnes  ME,  et  al.  Restoration  of  metabolic  health  by  decreased  consumption  of  branched‐chain  amino  acids.  J  Physiol. 2018; 596:623–45.     https://doi.org/10.1113/JP275075 

42.  Levothyroxine enhances glucose clearance and blunts  the onset of experimental type 1 diabetes mellitus in  mice. Br J Pharmacol. 2017; 174:3795–810.     https://doi.org/10.1111/bph.13975 

33.  Fontana  L,  Cummings  NE,  Arriola  Apelo  SI,  Neuman  JC, Kasza I, Schmidt BA, Cava E, Spelta F, Tosti V, Syed  FA, Baar EL, Veronese N, Cottrell SE, et al. Decreased  Consumption  of  Branched‐Chain  Amino  Acids  Improves  Metabolic  Health.  Cell  Reports.  2016;  16:520–30.  https://doi.org/10.1016/j.celrep.2016.05.092 

43.  Ozerov  IV,  Lezhnina  KV,  Izumchenko  E,  Artemov  AV,  Medintsev S, Vanhaelen Q, Aliper A, Vijg J, Osipov AN,  Labat I, West MD, Buzdin A, Cantor CR, et al. In silico  Pathway  Activation  Network  Decomposition  Analysis  (iPANDA)  as  a  method  for  biomarker  development.  Nat Commun. 2016; 7:13427.     https://doi.org/10.1038/ncomms13427 

+ 34.  Verdin  E.  NAD   in  aging,  metabolism,  and  neuro‐ degeneration. Science. 2015; 350:1208–13.     https://doi.org/10.1126/science.aac4854 

44.  Olshansky  SJ.  From  Lifespan  to  Healthspan.  JAMA.  2018; 320:1323–24.     https://doi.org/10.1001/jama.2018.12621 

35.  Yoshino  J,  Baur  JA,  Imai  SI.  NAD+  Intermediates:  The  Biology  and  Therapeutic  Potential  of  NMN  and  NR.  Cell Metab. 2018; 27:513–28.     https://doi.org/10.1016/j.cmet.2017.11.002 

45.  Aliper  A,  Plis  S,  Artemov  A,  Ulloa  A,  Mamoshina  P,  Zhavoronkov  A.  Deep  Learning  Applications  for  Predicting  Pharmacological  Properties  of  Drugs  and  Drug Repurposing Using Transcriptomic Data. Mol  

www.aging‐us.com 



AGING

 

Pharm. 2016; 13:2524–30.   https://doi.org/10.1021/acs.molpharmaceut.6b00248 

https://doi.org/10.1016/j.ebiom.2017.04.013  56.  Kennedy BK, Lamming DW. The Mechanistic Target of  Rapamycin:  The  Grand  ConducTOR  of  Metabolism  and Aging. Cell Metab. 2016; 23:990–1003.     https://doi.org/10.1016/j.cmet.2016.05.009 

46.  Jellen  LC,  Aliper  A,  Buzdin  A,  Zhavoronkov  A.  Screening  and  personalizing  nootropic  drugs  and  cognitive  modulator  regimens  in  silico.  Front  Syst  Neurosci. 2015; 9:4.     https://doi.org/10.3389/fnsys.2015.00004 

57.  Elhassan  YS,  Philp  AA,  Lavery  GG.  Targeting  NAD+  in  Metabolic  Disease:  New  Insights  Into  an  Old  Molecule. J Endocr Soc. 2017; 1:816–35.     https://doi.org/10.1210/js.2017‐00092 

47.  Mamoshina  P,  Vieira  A,  Putin  E,  Zhavoronkov  A.  Applications  of  Deep  Learning  in  Biomedicine.  Mol  Pharm. 2016; 13:1445–54.     https://doi.org/10.1021/acs.molpharmaceut.5b00982 

58.  Fang EF, Lautrup S, Hou Y, Demarest TG, Croteau DL,  Mattson  MP,  Bohr  VA.  NAD+  in  Aging:  Molecular  Mechanisms  and  Translational  Implications.  Trends  Mol Med. 2017; 23:899–916.     https://doi.org/10.1016/j.molmed.2017.08.001 

48.  Aliper A, Belikov AV, Garazha A, Jellen L, Artemov A,  Suntsova M, Ivanova A, Venkova L, Borisov N, Buzdin  A, Mamoshina P, Putin E, Swick AG, et al. In search for  geroprotectors:  in  silico  screening  and  in  vitro  validation  of  signalome‐level  mimetics  of  young  healthy  state.  Aging  (Albany  NY).  2016;  8:2127–52.  https://doi.org/10.18632/aging.101047 

59.  Oh  J,  Lee  YD,  Wagers  AJ.  Stem  cell  aging:  mechanisms,  regulators  and  therapeutic  opportunities. Nat Med. 2014; 20:870–80.     https://doi.org/10.1038/nm.3651 

49.  Aliper  A,  Jellen  L,  Cortese  F,  Artemov  A,  Karpinsky‐ Semper  D,  Moskalev  A,  Swick  AG,  Zhavoronkov  A.  Towards  natural  mimetics  of  metformin  and  rapamycin.  Aging  (Albany  NY).  2017;  9:2245–68.  https://doi.org/10.18632/aging.101319 

60.  Jiang  F,  Jiang  Y,  Zhi  H,  Dong  Y,  Li  H,  Ma  S,  Wang  Y,  Dong  Q,  Shen  H,  Wang  Y.  Artificial  intelligence  in  healthcare:  past,  present  and  future.  Stroke  Vasc  Neurol. 2017; 2:230–43. https://doi.org/10.1136/svn‐ 2017‐000101 

50.  Barardo DG, Newby D, Thornton D, Ghafourian T, de  Magalhães  JP,  Freitas  AA.  Machine  learning  for  predicting  lifespan‐extending  chemical  compounds.  Aging (Albany NY). 2017; 9:1721–37.     https://doi.org/10.18632/aging.101264 

61.  Mamoshina  P,  Ojomoko  L,  Yanovich  Y,  Ostrovski  A,  Botezatu  A,  Prikhodko  P,  Izumchenko  E,  Aliper  A,  Romantsov  K,  Zhebrak  A,  Ogu  IO,  Zhavoronkov  A,  Mamoshina P, et al. Converging blockchain and next‐ generation  artificial  intelligence  technologies  to  decentralize  and  accelerate  biomedical  research  and  healthcare. Oncotarget. 2017; 9:5665–90.     https://doi.org/10.18632/oncotarget.22345 

51.  Dönertaş  HM,  Fuentealba  Valenzuela  M,  Partridge  L,  Thornton  JM.  Gene  expression‐based  drug  repurposing  to  target  aging.  Aging  Cell.  2018;  17:e12819. https://doi.org/10.1111/acel.12819 

62.  Peters  MJ,  Joehanes  R,  Pilling  LC,  Schurmann  C,  Conneely  KN,  Powell  J,  Reinmaa  E,  Sutphin  GL,  Zhernakova  A,  Schramm  K,  Wilson  YA,  Kobes  S,  Tukiainen  T,  et  al,  and  NABEC/UKBEC  Consortium.  The  transcriptional  landscape  of  age  in  human  peripheral  blood.  Nat  Commun.  2015;  6:8570.  https://doi.org/10.1038/ncomms9570 

52.  Fang  J,  Gao  L,  Ma  H,  Wu  Q,  Wu  T,  Wu  J,  Wang  Q,  Cheng  F.  Quantitative  and  Systems  Pharmacology  3.  Network‐Based  Identification  of  New  Targets  for  Natural  Products  Enables  Potential  Uses  in  Aging‐ Associated  Disorders.  Front  Pharmacol.  2017;  8:747.  https://doi.org/10.3389/fphar.2017.00747  53.  Hwang  HV,  Tran  DT,  Rebuffatti  MN,  Li  CS,  Knowlton  AA.  Investigation  of  quercetin  and  hyperoside  as  senolytics in adult human endothelial cells. PLoS One.  2018; 13:e0190374.     https://doi.org/10.1371/journal.pone.0190374 

63.  Emilsson V, Ilkov M, Lamb JR, Finkel N, Gudmundsson  EF, Pitts R, Hoover H, Gudmundsdottir V, Horman SR,  Aspelund T, Shu L, Trifonov V, Sigurdsson S, et al. Co‐ regulatory  networks  of  human  serum  proteins  link  genetics  to  disease.  Science.  2018;  361:769–73.  https://doi.org/10.1126/science.aaq1327 

54.  Xu  M,  Pirtskhalava  T,  Farr  JN,  Weigand  BM,  Palmer  AK, Weivoda MM, Inman CL, Ogrodnik MB, Hachfeld  CM, Fraser DG, Onken JL, Johnson KO, Verzosa GC, et  al.  Senolytics  improve  physical  function  and  increase  lifespan  in  old  age.  Nat  Med.  2018;  24:1246–56.  https://doi.org/10.1038/s41591‐018‐0092‐9 

64.  Mamoshina  P,  Kochetov  K,  Putin  E,  Cortese  F,  Aliper  A,  Lee  WS,  Ahn  SM,  Uhn  L,  Skjodt  N,  Kovalchuk  O,  Scheibye‐Knudsen  M,  Zhavoronkov  A.  Population  specific biomarkers of human aging: a big data study  using  South  Korean,  Canadian  and  Eastern  European  patient  populations.  J  Gerontol  A  Biol  Sci  Med  Sci.  2018; 73:1482–90.  

55.  Kirkland JL, Tchkonia T. Cellular Senescence: A Trans‐ lational Perspective. EBioMedicine. 2017; 21:21–28.  

www.aging‐us.com 



AGING

 

https://doi.org/10.1093/gerona/gly005 

65.  Putin  E,  Mamoshina  P,  Aliper  A,  Korzinkin  M,  Moskalev A, Kolosov A, Ostrovskiy A, Cantor C, Vijg J,  Zhavoronkov  A.  Deep  biomarkers  of  human  aging:  application  of  deep  neural  networks  to  biomarker  development.  Aging  (Albany  NY).  2016;  8:1021–33.  https://doi.org/10.18632/aging.100968   

www.aging‐us.com 

10 

AGING